1
|
Ahmed Zaki MS, Haidara MA, Abdallaa AM, Mohammed H, Sideeg AM, Eid RA. Role of dietary selenium in alleviating bisphenol A toxicity of liver albino rats: Histological, ultrastructural, and biomarker assessments. J Food Biochem 2021; 45:e13725. [PMID: 33847390 DOI: 10.1111/jfbc.13725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023]
Abstract
Bisphenol A (BPA) is used as a plasticizer in polycarbonate plastics. It has been used in consumer products and epoxy resins for decades as protective coatings and linings for food and beverage bottles. This can trigger human reactions to BPA which interferes with estrogen receptors. Our study explored the ameliorative effects of selenium (Se) in male rats on liver damage caused by BPA. Rats were divided into four groups at random: The first one obtained olive oil and acted as a control. Se (0.5 mg/kg diet) was given for the second group. The third one was treated with BPA (10 mg/kg body weight/day) orally. Concomitantly Se (0.5 mg/kg diet) and BPA (10 mg/kg body weight/day) were given orally in the fourth one. Liver specimens were prepared for light, electron microscopes and the serum samples were screened for biochemical markers. In the BPA received group, histological findings indicated apoptotic hepatic histological changes such as sinusoidal congestion, cytoplasmic vacuolation and leukocyte infiltration. Ultrastructurally, the same group had mitochondrial degeneration, rough endoplasmic reticulum swelling, and nuclear pyknosis, as well as fat droplet deposition and lysosome enhancement. Liver enzymes: In the BPA group, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) have been substantially increased. Moreover, histological and ultrastructural improvements were seen in the rat population treated with BPA and Se, whereas ALT and AST levels were lowered and malondialdehyde (MDA), glutathione peroxidase (GPx), human C reactive protein (hCRP), and the serum levels of interleukin-6 (IL-6) were significantly modulated. PRACTICAL APPLICATIONS: Bisphenol A (BPA) is used in the manufacturing of polycarbonate plastic (e.g., water bottles, baby bottles) and epoxy resins (e.g., inner coating in metallic food cans). It is a non-polymer preservative for other plastics, one of the contaminants of the atmosphere and a common endocrine estrogenic disruptor. Our study explored the ameliorative effects of selenium (Se) in male rats on liver damage caused by BPA. Rats were divided into four groups at random: The first one obtained olive oil and acted as a control. Se (0.5 mg/kg diet) was given for the second group. The third one was treated with BPA (10 mg/kg body weight/day) orally. Concomitant Se (0.5 mg/kg diet) and BPA (10 mg/kg body weight/day) were given in the fourth one. Liver specimens were prepared for light, electron microscopes and the serum samples were screened for biochemical markers.
Collapse
Affiliation(s)
- Mohamed Samir Ahmed Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha, Saudi Arabia.,Histology Department, College of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed A Haidara
- Physiology Department, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Asim M Abdallaa
- Anatomy Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Heitham Mohammed
- Anatomy Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abulqasim M Sideeg
- Anatomy Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Refaat A Eid
- Pathology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
2
|
Hu DD, Zhao Q, Cheng Y, Xiao XR, Huang JF, Qu Y, Li X, Tang YM, Bao WM, Yang JH, Jiang T, Hu JP, Gonzalez FJ, Li F. The Protective Roles of PPARα Activation in Triptolide-Induced Liver Injury. Toxicol Sci 2019; 171:1-12. [PMID: 31241159 PMCID: PMC11514144 DOI: 10.1093/toxsci/kfz146] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Triptolide (TP), one of the main active ingredients in Tripterygium wilfordii Hook F, is clinically used to treat immune diseases but is known to cause liver injury. The aim of this study was to investigate the biomarkers for TP-induced hepatotoxicity in mice and to determine potential mechanisms of its liver injury. LC/MS-based metabolomics was used to determine the metabolites that were changed in TP-induced liver injury. The accumulation of long-chain acylcarnitines in serum indicated that TP exposure disrupted endogenous peroxisome proliferator-activated receptor α (PPARα) signaling. Triptolide-induced liver injury could be alleviated by treatment of mice with the PPARα agonist fenofibrate, whereas the PPARα antagonist GW6471 increased hepatotoxicity. Furthermore, fenofibrate did not protect Ppara-/- mice from TP-induced liver injury, suggesting an essential role for the PPARα in the protective effect of fenofibrate. Elevated long-chain acylcarnitines may protect TP-induced liver injury through activation of the NOTCH-NRF2 pathway as revealed in primary mouse hepatocytes and in vivo. In agreement with these observations in mice, the increase in long-chain acylcarnitines was observed in the serum of patients with cholestatic liver injury compared with healthy volunteers. These data demonstrated the role of PPARα and long-chain acylcarnitines in TP-induced hepatotoxicity, and suggested that modulation of PPARα may protect against drug-induced liver injury.
Collapse
Affiliation(s)
- Dan-Dan Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University, Kunming 650500, China
| | - Qi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yan Cheng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xue-Rong Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Jian-Feng Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yan Qu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xian Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University, Kunming 650500, China
| | - Ying-Mei Tang
- Department of Gastroenterology, Yunnan Research Center for Liver Diseases, The 2nd Affiliated Hospital of Kunming Medical University, Kunming 650033, China
| | - Wei-Min Bao
- Department of General Surgery, Yunnan Provincial 1st People’s Hospital, Kunming 650032, China
| | - Jin-Hui Yang
- Department of Gastroenterology, Yunnan Research Center for Liver Diseases, The 2nd Affiliated Hospital of Kunming Medical University, Kunming 650033, China
| | - Tao Jiang
- Department of Gastroenterology, Yunnan Research Center for Liver Diseases, The 2nd Affiliated Hospital of Kunming Medical University, Kunming 650033, China
| | - Jia-Peng Hu
- Clinical Laboratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming 650033, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Center for Cancer Research, Bethesda, Maryland 20892
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
3
|
Mahmoudi A, Hadrich F, Feki I, Ghorbel H, Bouallagui Z, Marrekchi R, Fourati H, Sayadi S. Oleuropein and hydroxytyrosol rich extracts from olive leaves attenuate liver injury and lipid metabolism disturbance in bisphenol A-treated rats. Food Funct 2018; 9:3220-3234. [PMID: 29781491 DOI: 10.1039/c8fo00248g] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the present study, we investigated the protective effects of oleuropein- and hydroxytyrosol-rich extracts obtained from olive leaves against bisphenol A (BPA)-induced hyperlipidemia and liver injury in male rats. For this purpose, four groups of male rats (8 per group) were used: control group (Control), rats treated with BPA, rats treated with both BPA and oleuropein (OLE-BPA), and rats treated with both BPA and hydroxytyrosol (HYT-BPA). After 60 days of treatment, the results obtained using the DXA technique showed that treatment with BPA (10 mg per kg b.w.) increased the body weight and adipose tissue mass in male rats. Moreover, plasma levels of triglycerides, total cholesterol, LDL-cholesterol, AST, ALT, LDH, and TNF-α increased. The immunohistochemical analysis revealed a significant increase in the expression of COX-2 and p53 and a decrease in the expression of Bcl-2 related to liver inflammation. Oral administration of oleuropein and hydroxytyrosol-rich extracts obtained from olive leaves at 16 mg kg-1 reduced both the body weight and adipose tissue mass. These extracts were able to ameliorate liver damage and improve the elevated levels of TG and liver enzymes of BPA-treated rats possibly through enhancing CAT and SOD activities. Western blot results revealed that administration of the abovementioned extracts decreased the protein expression of NF-κB and TNF-α through the p38 signaling pathway. Overall, the findings suggest that the olive leaf extracts possess hypolipidemic and hepatoprotective effects against BPA-induced metabolic disorders through enhancing the antioxidative defense system and regulating the important signaling pathway activities.
Collapse
Affiliation(s)
- Asma Mahmoudi
- Environmental Bioprocesses Laboratory, Center of Biotechnology of Sfax, University of Sfax, P.O.Box 1177, 3038 Sfax, Tunisia.
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Nikravesh H, Khodayar MJ, Mahdavinia M, Mansouri E, Zeidooni L, Dehbashi F. Protective Effect of Gemfibrozil on Hepatotoxicity Induced by Acetaminophen in Mice: the Importance of Oxidative Stress Suppression. Adv Pharm Bull 2018; 8:331-339. [PMID: 30023335 PMCID: PMC6046434 DOI: 10.15171/apb.2018.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 03/10/2018] [Accepted: 04/08/2018] [Indexed: 12/24/2022] Open
Abstract
Purpose: Gemfibrozil (GEM) apart from agonist activity at peroxisome proliferator-activated receptor-alpha (PPAR-α) has antioxidant and anti-inflammatory properties. Accordingly, the present study was designed to investigate the protective effect of GEM on acute liver toxicity induced by acetaminophen (APAP) in mice. Methods: In this study, mice divided in seven groups include, control group, APAP group, GEM group, three APAP groups pretreated with GEM at the doses of 25, 50 and 100 mg/kg respectively and APAP group pretreated with N-Acetyl cysteine. GEM, NAC or vehicle were administered for 10 days. In last day, GEM and NAC were gavaged 1 h before and 1 h after APAP injection. Twenty four hours after APAP, mice were sacrificed. Serum parameters include alanine aminotransferase (ALT), aspartate aminotransferase (AST) and liver tissue markers including catalase enzyme activity, reactive oxygen species (ROS), malondialdehyde and reduced glutathione (GSH) levels determined and histopathological parameters measured. Results: GEM led to significant decrease in serum ALT and AST activities and increase in catalase activity and hepatic GSH level and reduces malondialdehyde and ROS levels in the liver tissue. In confirmation, histopathological findings revealed that GEM decrease degeneration, vacuolation and necrosis of hepatocytes and infiltration of inflammatory cells. Conclusion: Present data demonstrated that GEM has antioxidant properties and can protect the liver from APAP toxicity, just in the same pathway that toxicity occurs by toxic ROS and that GEM may be an alternative therapeutic agent to NAC in APAP toxicity.
Collapse
Affiliation(s)
- Hojatolla Nikravesh
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Zeidooni
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fereshteh Dehbashi
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
5
|
Sukhotnik I, Nissimov N, Ben Shahar Y, Moati D, Bitterman N, Pollak Y, Berkowitz D, Coran AG, Bitterman A. Fenofibrate reduces intestinal damage and improves intestinal recovery following intestinal ischemia-reperfusion injury in a rat. Pediatr Surg Int 2016; 32:1193-1200. [PMID: 27651374 DOI: 10.1007/s00383-016-3979-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2016] [Indexed: 12/28/2022]
Abstract
PURPOSE Fenofibrate (FEN) is known as a nuclear receptor activator which regulates many pathophysiological processes, such as oxidative stress, inflammation, and leukocyte endothelium interactions. Recent studies have demonstrated an anti-oxidant, anti-inflammatory, and anti-ischemic role of FEN in the attenuation of ischemia-reperfusion (IR) injury in the kidney, liver, brain, and heart. The purpose of the present study was to examine the effect of FEN on intestinal recovery and enterocyte turnover after intestinal IR injury in rats. METHODS Male Sprague-Dawley rats were divided into four experimental groups: (1) sham rats underwent laparotomy, (2) sham-FEN rats underwent laparotomy and were treated with intraperitoneal (IP) FEN (20 mg/kg); (3) IR rats underwent occlusion of both the superior mesenteric artery and the portal vein for 30 min followed by 24 h of reperfusion, and (4) IR-FEN rats underwent IR and were treated with IP FEN immediately before abdominal closure. Intestinal structural changes, Park's injury score, enterocyte proliferation, and enterocyte apoptosis were determined 24 h following IR. The expression of Bax, Bcl-2, p-ERK, and caspase-3 in the intestinal mucosa was determined using real-time PCR, Western blot, and immunohistochemistry. RESULTS Treatment with FEN resulted in a significant decrease in Park's injury score in jejunum (32 %) and ileum (33 %) compared to IR animals. IR-FEN rats also demonstrated a significant increase in mucosal weight in jejunum (23 %) and ileum (22 %), mucosal DNA (38 %) and protein (65 %) in jejunum, villus height in jejunum (17 %) and ileum (21 %), and crypt depth in ileum (14 %) compared to IR animals. IR-FEN rats also experienced significant proliferation rates as well as lower apoptotic indices in jejunum and ileum which was accompanied with higher Bcl-2 levels compared to IR animals. CONCLUSIONS Treatment with fenofibrate prevents intestinal mucosal damage and stimulates intestinal epithelial cell turnover following intestinal IR in a rat model.
Collapse
Affiliation(s)
- I Sukhotnik
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel. .,Department of Pediatric Surgery B, Bnai Zion Medical Center, 47 Golomb St., P.O.B. 4940, 31048, Haifa, Israel.
| | - N Nissimov
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Pediatric Surgery B, Bnai Zion Medical Center, 47 Golomb St., P.O.B. 4940, 31048, Haifa, Israel
| | - Y Ben Shahar
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Surgery, Carmel Medical Center, Haifa, Israel
| | - D Moati
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - N Bitterman
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Y Pollak
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - D Berkowitz
- Department of Pediatric Gastroenterology, Bnai Zion Medical Center, Haifa, Israel
| | - A G Coran
- Section of Pediatric Surgery, C.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, MI, USA
| | - A Bitterman
- Department of Surgery, Carmel Medical Center, Haifa, Israel
| |
Collapse
|
6
|
Protective effects of fenofibrate against acute lung injury induced by intestinal ischemia/reperfusion in mice. Sci Rep 2016; 6:22044. [PMID: 26902261 PMCID: PMC4763198 DOI: 10.1038/srep22044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/04/2016] [Indexed: 11/08/2022] Open
Abstract
This experiment was conducted to evaluate whether pretreatment with fenofibrate could mitigate acute lung injury (ALI) in a mice model of intestinal ischemia/reperfusion (I/R). Male C57BL/6 mice were randomly assigned into three groups (n = 6): sham, intestinal I/R + vehicle, and intestinal I/R + fenofibrate. Intestinal I/R was achieved by clamping the superior mesenteric artery. Fenofibrate (100 mg/kg) or equal volume of vehicle was injected intraperitoneally 60 minutes before the ischemia. At the end of experiment, measurement of pathohistological score, inflammatory mediators and other markers were performed. In addition, a 24-hour survival experiment was conducted in intestinal I/R mice treated with fenofibrate or vehicle. The chief results were as anticipated. Pathohistological evaluation indicated that fenofibrate ameliorated the local intestine damage and distant lung injury. Pretreatment with fenofibrate significantly decreased inflammatory factors in both the intestine and the lung. Consistently, renal creatine levels and hepatic ALT levels were significantly decreased in the fenofibrate group. Moreover, serum systemic inflammatory response indicators were significantly alleviated in the fenofibrate group. In addition, fenofibrate administration significantly improved the survival rate. Collectively, our data indicated that pretreatment with fenofibrate prior to ischemia attenuated intestinal I/R injury and ALI.
Collapse
|
7
|
Lv J, Wang X, Liu S, Liang P, Feng M, Zhang L, Xu A. Protective effect of Fenofibrate in renal ischemia reperfusion injury: Involved in suppressing kinase 2 (JAK2)/transcription 3 (STAT3)/p53 signaling activation. ACTA ACUST UNITED AC 2015; 63:236-42. [DOI: 10.1016/j.patbio.2015.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/30/2015] [Indexed: 12/14/2022]
|
8
|
Cámara-Lemarroy CR, Guzman-DE LA Garza FJ, Cordero-Perez P, Ibarra-Hernandez JM, Muñoz-Espinosa LE, Fernandez-Garza NE. Gemfibrozil attenuates the inflammatory response and protects rats from abdominal sepsis. Exp Ther Med 2015; 9:1018-1022. [PMID: 25667670 PMCID: PMC4316892 DOI: 10.3892/etm.2015.2190] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 11/28/2014] [Indexed: 01/05/2023] Open
Abstract
Sepsis is a serious condition characterized by an infectious process that induces a severe systemic inflammatory response. In this study, the effects of gemfibrozil (GFZ) on the inflammatory response associated with abdominal sepsis were investigated using a rat model of cecal-ligation and puncture (CLP). Male Wistar rats were randomly divided into three groups: Sham-operated group (sham), where laparotomy was performed, the intestines were manipulated, and the cecum was ligated but not punctured; control group, subjected to CLP; and GFZ group, which received GFZ prior to undergoing CLP. The groups were then subdivided into three different time-points: 2, 4 and 24 h, indicating the time at which blood samples were obtained for analysis. Serum concentrations of tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), malondialdehyde (MDA), aspartate aminotransferase (AST), alanine aminotransferase (ALT) and lactate dehydrogenase (LDH) were determined. The LDH, AST and ALT values were significantly elevated following CLP compared with those in the sham group, and GFZ treatment was able to reduce these elevations. GFZ also reduced the sepsis-induced elevations of TNF-α and IL-1. In conclusion, GFZ treatment was able to attenuate the inflammatory response associated with CLP-induced sepsis, by diminishing the release of inflammatory cytokines, thereby reducing tissue injury and oxidative stress.
Collapse
Affiliation(s)
- Carlos R Cámara-Lemarroy
- Department of Internal Medicine, University Hospital 'José Eleuterio González', Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| | | | - Paula Cordero-Perez
- Liver Unit, Department of Internal Medicine, University Hospital 'José Eleuterio González', Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| | - Juan M Ibarra-Hernandez
- Department of Physiology, School of Medicine, Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| | - Linda E Muñoz-Espinosa
- Liver Unit, Department of Internal Medicine, University Hospital 'José Eleuterio González', Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| | - Nancy E Fernandez-Garza
- Department of Physiology, School of Medicine, Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| |
Collapse
|
9
|
N-acetyl-serotonin protects HepG2 cells from oxidative stress injury induced by hydrogen peroxide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:310504. [PMID: 25013541 PMCID: PMC4074966 DOI: 10.1155/2014/310504] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 04/10/2014] [Accepted: 05/03/2014] [Indexed: 12/12/2022]
Abstract
Oxidative stress plays an important role in the pathogenesis of liver diseases. N-Acetyl-serotonin (NAS) has been reported to protect against oxidative damage, though the mechanisms by which NAS protects hepatocytes from oxidative stress remain unknown. To determine whether pretreatment with NAS could reduce hydrogen peroxide- (H2O2-) induced oxidative stress in HepG2 cells by inhibiting the mitochondrial apoptosis pathway, we investigated the H2O2-induced oxidative damage to HepG2 cells with or without NAS using MTT, Hoechst 33342, rhodamine 123, Terminal dUTP Nick End Labeling Assay (TUNEL), dihydrodichlorofluorescein (H2DCF), Annexin V and propidium iodide (PI) double staining, immunocytochemistry, and western blot. H2O2 produced dramatic injuries in HepG2 cells, represented by classical morphological changes of apoptosis, increased levels of malondialdehyde (MDA) and intracellular reactive oxygen species (ROS), decreased activity of superoxide dismutase (SOD), and increased activities of caspase-9 and caspase-3, release of cytochrome c (Cyt-C) and apoptosis-inducing factor (AIF) from mitochondria, and loss of membrane potential (ΔΨm). NAS significantly inhibited H2O2-induced changes, indicating that it protected against H2O2-induced oxidative damage by reducing MDA levels and increasing SOD activity and that it protected the HepG2 cells from apoptosis through regulating the mitochondrial apoptosis pathway, involving inhibition of mitochondrial hyperpolarization, release of mitochondrial apoptogenic factors, and caspase activity.
Collapse
|
10
|
Peroxisome Proliferator Activator Receptor (PPAR)- γ Ligand, but Not PPAR- α , Ameliorates Cyclophosphamide-Induced Oxidative Stress and Inflammation in Rat Liver. PPAR Res 2014; 2014:626319. [PMID: 24803924 PMCID: PMC3996363 DOI: 10.1155/2014/626319] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 03/09/2014] [Accepted: 03/10/2014] [Indexed: 12/12/2022] Open
Abstract
Hepatoprotective potential of peroxisome proliferator activator receptor (PPAR)-α and -γ agonists, fenofibrate (FEN), and pioglitazone (PIO), respectively, against cyclophosphamide (CP)-induced toxicity has been investigated in rat. FEN and PIO (150 and 10 mg/kg/day, resp.) were given orally for 4 weeks. In separate groups, CP (150 mg/kg, i.p.) was injected as a single dose 5 days before the end of experiment, with or without either PPAR agonist. CP induced hepatotoxicity, as it caused histopathological alterations, with increased serum alanine and aspartate transaminases, total bilirubin, albumin, alkaline phosphatase and lactate dehydrogenase. CP caused hepatic oxidative stress, indicated by decrease in tissue reduced glutathione, with increase in malondialdehyde and nitric oxide levels. CP also caused decrease in hepatic antioxidant enzyme levels, including catalase, superoxide dismutase, glutathione peroxidase, and glutathione S-transferase. Furthermore, CP increased serum and hepatic levels of the inflammatory marker tumor necrosis factor (TNF)-α, evaluated using ELISA. Preadministration of PIO, but not FEN, prior to CP challenge improved hepatic function and histology, and significantly reversed oxidative and inflammatory parameters. In conclusion, activation of PPAR-γ, but not PPAR-α, conferred protection against CP-induced hepatotoxicity, via activation of antioxidant and anti-inflammatory mechanisms, and may serve as supplement during CP chemotherapy.
Collapse
|
11
|
Sun J, Sun L, Zhang N, Lu X, Zhang H. A20 is up-regulated in primary mouse hepatocytes subjected to hypoxia and reperfusion. Cell Biochem Funct 2012; 30:683-6. [DOI: 10.1002/cbf.2850] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/22/2012] [Accepted: 05/17/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Jiao Sun
- Department of Pathophysiology, College of Basic Medical Sciences; China Medical University; Shenyang; China
| | - Luning Sun
- Department of Pathophysiology, College of Basic Medical Sciences; China Medical University; Shenyang; China
| | - Ning Zhang
- Department of Pathophysiology, College of Basic Medical Sciences; China Medical University; Shenyang; China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences; China Medical University; Shenyang; China
| | - Haipeng Zhang
- Department of Pathophysiology, College of Basic Medical Sciences; China Medical University; Shenyang; China
| |
Collapse
|